Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 17.440
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(14): 3585-3601.e22, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38821050

RESUMEN

Dolichol is a lipid critical for N-glycosylation as a carrier for activated sugars and nascent oligosaccharides. It is commonly thought to be directly produced from polyprenol by the enzyme SRD5A3. Instead, we found that dolichol synthesis requires a three-step detour involving additional metabolites, where SRD5A3 catalyzes only the second reaction. The first and third steps are performed by DHRSX, whose gene resides on the pseudoautosomal regions of the X and Y chromosomes. Accordingly, we report a pseudoautosomal-recessive disease presenting as a congenital disorder of glycosylation in patients with missense variants in DHRSX (DHRSX-CDG). Of note, DHRSX has a unique dual substrate and cofactor specificity, allowing it to act as a NAD+-dependent dehydrogenase and as a NADPH-dependent reductase in two non-consecutive steps. Thus, our work reveals unexpected complexity in the terminal steps of dolichol biosynthesis. Furthermore, we provide insights into the mechanism by which dolichol metabolism defects contribute to disease.


Asunto(s)
Dolicoles , Dolicoles/metabolismo , Dolicoles/biosíntesis , Humanos , Glicosilación , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Trastornos Congénitos de Glicosilación/metabolismo , Trastornos Congénitos de Glicosilación/genética , Masculino , Mutación Missense , Femenino
2.
Cell ; 186(10): 2193-2207.e19, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37098343

RESUMEN

Somatic hypermutation (SHM), initiated by activation-induced cytidine deaminase (AID), generates mutations in the antibody-coding sequence to allow affinity maturation. Why these mutations intrinsically focus on the three nonconsecutive complementarity-determining regions (CDRs) remains enigmatic. Here, we found that predisposition mutagenesis depends on the single-strand (ss) DNA substrate flexibility determined by the mesoscale sequence surrounding AID deaminase motifs. Mesoscale DNA sequences containing flexible pyrimidine-pyrimidine bases bind effectively to the positively charged surface patches of AID, resulting in preferential deamination activities. The CDR hypermutability is mimicable in in vitro deaminase assays and is evolutionarily conserved among species using SHM as a major diversification strategy. We demonstrated that mesoscale sequence alterations tune the in vivo mutability and promote mutations in an otherwise cold region in mice. Our results show a non-coding role of antibody-coding sequence in directing hypermutation, paving the way for the synthetic design of humanized animal models for optimal antibody discovery and explaining the AID mutagenesis pattern in lymphoma.


Asunto(s)
Citidina Desaminasa , Hipermutación Somática de Inmunoglobulina , Animales , Ratones , Anticuerpos/genética , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , ADN/genética , ADN de Cadena Simple , Mutación , Evolución Molecular , Regiones Determinantes de Complementariedad/genética , Motivos de Nucleótidos
3.
Cell ; 185(6): 1065-1081.e23, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35245431

RESUMEN

Motor behaviors are often planned long before execution but only released after specific sensory events. Planning and execution are each associated with distinct patterns of motor cortex activity. Key questions are how these dynamic activity patterns are generated and how they relate to behavior. Here, we investigate the multi-regional neural circuits that link an auditory "Go cue" and the transition from planning to execution of directional licking. Ascending glutamatergic neurons in the midbrain reticular and pedunculopontine nuclei show short latency and phasic changes in spike rate that are selective for the Go cue. This signal is transmitted via the thalamus to the motor cortex, where it triggers a rapid reorganization of motor cortex state from planning-related activity to a motor command, which in turn drives appropriate movement. Our studies show how midbrain can control cortical dynamics via the thalamus for rapid and precise motor behavior.


Asunto(s)
Corteza Motora , Movimiento , Tálamo , Animales , Mesencéfalo , Ratones , Corteza Motora/fisiología , Neuronas/fisiología , Tálamo/fisiología
4.
Cell ; 184(14): 3762-3773.e10, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34133943

RESUMEN

Sneezing is a vital respiratory reflex frequently associated with allergic rhinitis and viral respiratory infections. However, its neural circuit remains largely unknown. A sneeze-evoking region was discovered in both cat and human brainstems, corresponding anatomically to the central recipient zone of nasal sensory neurons. Therefore, we hypothesized that a neuronal population postsynaptic to nasal sensory neurons mediates sneezing in this region. By screening major presynaptic neurotransmitters/neuropeptides released by nasal sensory neurons, we found that neuromedin B (NMB) peptide is essential for signaling sneezing. Ablation of NMB-sensitive postsynaptic neurons in the sneeze-evoking region or deficiency in NMB receptor abolished the sneezing reflex. Remarkably, NMB-sensitive neurons further project to the caudal ventral respiratory group (cVRG). Chemical activation of NMB-sensitive neurons elicits action potentials in cVRG neurons and leads to sneezing behavior. Our study delineates a peptidergic pathway mediating sneezing, providing molecular insights into the sneezing reflex arc.


Asunto(s)
Tronco Encefálico/fisiopatología , Neuropéptidos/metabolismo , Nariz/fisiopatología , Reflejo/fisiología , Estornudo/fisiología , Animales , Modelos Animales de Enfermedad , Hipersensibilidad/fisiopatología , Masculino , Ratones Endogámicos C57BL , Neuroquinina B/análogos & derivados , Neuroquinina B/metabolismo , Neuronas/metabolismo , ARN Interferente Pequeño/metabolismo , Células Receptoras Sensoriales/fisiología , Canales Catiónicos TRPV/metabolismo , Grabación en Video
5.
Cell ; 184(18): 4680-4696.e22, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34380047

RESUMEN

Mutations causing amyotrophic lateral sclerosis (ALS) often affect the condensation properties of RNA-binding proteins (RBPs). However, the role of RBP condensation in the specificity and function of protein-RNA complexes remains unclear. We created a series of TDP-43 C-terminal domain (CTD) variants that exhibited a gradient of low to high condensation propensity, as observed in vitro and by nuclear mobility and foci formation. Notably, a capacity for condensation was required for efficient TDP-43 assembly on subsets of RNA-binding regions, which contain unusually long clusters of motifs of characteristic types and density. These "binding-region condensates" are promoted by homomeric CTD-driven interactions and required for efficient regulation of a subset of bound transcripts, including autoregulation of TDP-43 mRNA. We establish that RBP condensation can occur in a binding-region-specific manner to selectively modulate transcriptome-wide RNA regulation, which has implications for remodeling RNA networks in the context of signaling, disease, and evolution.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Regiones no Traducidas 3'/genética , Secuencia de Bases , Núcleo Celular/metabolismo , Células HEK293 , Células HeLa , Homeostasis , Humanos , Mutación/genética , Motivos de Nucleótidos/genética , Transición de Fase , Mutación Puntual/genética , Poli A/metabolismo , Unión Proteica , Multimerización de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Eliminación de Secuencia
6.
Cell ; 184(17): 4564-4578.e18, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34302739

RESUMEN

The mesencephalic locomotor region (MLR) is a key midbrain center with roles in locomotion. Despite extensive studies and clinical trials aimed at therapy-resistant Parkinson's disease (PD), debate on its function remains. Here, we reveal the existence of functionally diverse neuronal populations with distinct roles in control of body movements. We identify two spatially intermingled glutamatergic populations separable by axonal projections, mouse genetics, neuronal activity profiles, and motor functions. Most spinally projecting MLR neurons encoded the full-body behavior rearing. Loss- and gain-of-function optogenetic perturbation experiments establish a function for these neurons in controlling body extension. In contrast, Rbp4-transgene-positive MLR neurons project in an ascending direction to basal ganglia, preferentially encode the forelimb behaviors handling and grooming, and exhibit a role in modulating movement. Thus, the MLR contains glutamatergic neuronal subpopulations stratified by projection target exhibiting roles in action control not restricted to locomotion.


Asunto(s)
Locomoción/fisiología , Mesencéfalo/anatomía & histología , Animales , Ganglios Basales/metabolismo , Conducta Animal , Femenino , Integrasas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Optogenética , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Médula Espinal/metabolismo , Transgenes , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
7.
Cell ; 184(9): 2362-2371.e9, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33735608

RESUMEN

The 501Y.V2 variants of SARS-CoV-2 containing multiple mutations in spike are now dominant in South Africa and are rapidly spreading to other countries. Here, experiments with 18 pseudotyped viruses showed that the 501Y.V2 variants do not confer increased infectivity in multiple cell types except for murine ACE2-overexpressing cells, where a substantial increase in infectivity was observed. Notably, the susceptibility of the 501Y.V2 variants to 12 of 17 neutralizing monoclonal antibodies was substantially diminished, and the neutralization ability of the sera from convalescent patients and immunized mice was also reduced for these variants. The neutralization resistance was mainly caused by E484K and N501Y mutations in the receptor-binding domain of spike. The enhanced infectivity in murine ACE2-overexpressing cells suggests the possibility of spillover of the 501Y.V2 variants to mice. Moreover, the neutralization resistance we detected for the 501Y.V2 variants suggests the potential for compromised efficacy of monoclonal antibodies and vaccines.


Asunto(s)
COVID-19/inmunología , COVID-19/virología , Evasión Inmune , SARS-CoV-2/patogenicidad , Enzima Convertidora de Angiotensina 2/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Línea Celular Tumoral , Células HEK293 , Humanos , Mutación/genética , SARS-CoV-2/genética
8.
Cell ; 181(5): 1062-1079.e30, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32386547

RESUMEN

Expansions of amino acid repeats occur in >20 inherited human disorders, and many occur in intrinsically disordered regions (IDRs) of transcription factors (TFs). Such diseases are associated with protein aggregation, but the contribution of aggregates to pathology has been controversial. Here, we report that alanine repeat expansions in the HOXD13 TF, which cause hereditary synpolydactyly in humans, alter its phase separation capacity and its capacity to co-condense with transcriptional co-activators. HOXD13 repeat expansions perturb the composition of HOXD13-containing condensates in vitro and in vivo and alter the transcriptional program in a cell-specific manner in a mouse model of synpolydactyly. Disease-associated repeat expansions in other TFs (HOXA13, RUNX2, and TBP) were similarly found to alter their phase separation. These results suggest that unblending of transcriptional condensates may underlie human pathologies. We present a molecular classification of TF IDRs, which provides a framework to dissect TF function in diseases associated with transcriptional dysregulation.


Asunto(s)
Expansión de las Repeticiones de ADN/genética , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Alanina/genética , Animales , Secuencia de Bases/genética , Expansión de las Repeticiones de ADN/fisiología , Modelos Animales de Enfermedad , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Mutación/genética , Linaje , Sindactilia/genética , Factores de Transcripción/metabolismo
9.
Cell ; 178(1): 190-201.e11, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31204101

RESUMEN

The placental transfer of maternal IgG is critical for infant protection against infectious pathogens. However, factors that modulate the placental transfer of IgG remain largely undefined. HIV-infected women have impaired placental IgG transfer, presenting a unique "disruption model" to define factors that modulate placental IgG transfer. We measured the placental transfer efficiency of maternal HIV and pathogen-specific IgG in US and Malawian HIV-infected mothers and their HIV-exposed uninfected and infected infants. We examined the role of maternal HIV disease progression, infant factors, placental Fc receptor expression, IgG subclass, and glycan signatures and their association with placental IgG transfer efficiency. Maternal IgG characteristics, such as binding to placentally expressed Fc receptors FcγRIIa and FcγRIIIa, and Fc region glycan profiles were associated with placental IgG transfer efficiency. Our findings suggest that Fc region characteristics modulate the selective placental transfer of IgG, with implications for maternal vaccine design and infant health.


Asunto(s)
Infecciones por VIH/transmisión , VIH/genética , Inmunoglobulina G/sangre , Transmisión Vertical de Enfermedad Infecciosa , Placenta/metabolismo , Complicaciones Infecciosas del Embarazo/virología , Receptores de IgG/metabolismo , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Glicosilación , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Fragmentos Fc de Inmunoglobulinas/metabolismo , Lactante , Recién Nacido , Malaui , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Estados Unidos , Carga Viral/genética
10.
Cell ; 177(3): 766-781.e24, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30955882

RESUMEN

During autophagy, vesicle dynamics and cargo recruitment are driven by numerous adaptors and receptors that become tethered to the phagophore through interactions with lipidated ATG8/LC3 decorating the expanding membrane. Most currently described ATG8-binding proteins exploit a well-defined ATG8-interacting motif (AIM, or LC3-interacting region [LIR]) that contacts a hydrophobic patch on ATG8 known as the LIR/AIM docking site (LDS). Here we describe a new class of ATG8 interactors that exploit ubiquitin-interacting motif (UIM)-like sequences for high-affinity binding to an alternative ATG8 interaction site. Assays with candidate UIM-containing proteins together with unbiased screens identified a large collection of UIM-based ATG8 interactors in plants, yeast, and humans. Analysis of a subset also harboring ubiquitin regulatory X (UBX) domains revealed a role for UIM-directed autophagy in clearing non-functional CDC48/p97 complexes, including some impaired in human disease. With this new class of adaptors and receptors, we greatly extend the reach of selective autophagy and identify new factors regulating autophagic vesicle dynamics.


Asunto(s)
Familia de las Proteínas 8 Relacionadas con la Autofagia/metabolismo , Autofagia , Proteínas Asociadas a Microtúbulos/metabolismo , Secuencias de Aminoácidos , Arabidopsis/metabolismo , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Familia de las Proteínas 8 Relacionadas con la Autofagia/química , Sitios de Unión , Humanos , Proteínas Asociadas a Microtúbulos/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia
11.
Immunity ; 57(6): 1394-1412.e8, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38821054

RESUMEN

Recent single-cell RNA sequencing studies have revealed distinct microglial states in development and disease. These include proliferative-region-associated microglia (PAMs) in developing white matter and disease-associated microglia (DAMs) prevalent in various neurodegenerative conditions. PAMs and DAMs share a similar core gene signature. However, the extent of the dynamism and plasticity of these microglial states, as well as their functional significance, remains elusive, partly due to the lack of specific tools. Here, we generated an inducible Cre driver line, Clec7a-CreERT2, that targets PAMs and DAMs in the brain parenchyma. Utilizing this tool, we profiled labeled cells during development and in several disease models, uncovering convergence and context-dependent differences in PAM and DAM gene expression. Through long-term tracking, we demonstrated microglial state plasticity. Lastly, we specifically depleted DAMs in demyelination, revealing their roles in disease recovery. Together, we provide a versatile genetic tool to characterize microglial states in CNS development and disease.


Asunto(s)
Plasticidad de la Célula , Microglía , Remielinización , Microglía/fisiología , Animales , Ratones , Plasticidad de la Célula/genética , Enfermedades Desmielinizantes/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales de Enfermedad , Encéfalo , Vaina de Mielina/metabolismo , Sustancia Blanca/patología
12.
Cell ; 174(1): 102-116.e14, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29804837

RESUMEN

RAG endonuclease initiates antibody heavy chain variable region exon assembly from V, D, and J segments within a chromosomal V(D)J recombination center (RC) by cleaving between paired gene segments and flanking recombination signal sequences (RSSs). The IGCR1 control region promotes DJH intermediate formation by isolating Ds, JHs, and RCs from upstream VHs in a chromatin loop anchored by CTCF-binding elements (CBEs). How VHs access the DJHRC for VH to DJH rearrangement was unknown. We report that CBEs immediately downstream of frequently rearranged VH-RSSs increase recombination potential of their associated VH far beyond that provided by RSSs alone. This CBE activity becomes particularly striking upon IGCR1 inactivation, which allows RAG, likely via loop extrusion, to linearly scan chromatin far upstream. VH-associated CBEs stabilize interactions of D-proximal VHs first encountered by the DJHRC during linear RAG scanning and thereby promote dominant rearrangement of these VHs by an unanticipated chromatin accessibility-enhancing CBE function.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Cromatina/metabolismo , Proteínas de Homeodominio/metabolismo , Recombinación V(D)J , Animales , Línea Celular , ADN Intergénico/genética , ADN Intergénico/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/metabolismo , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Mutagénesis , Señales de Clasificación de Proteína , ARN Guía de Kinetoplastida/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
13.
Cell ; 168(6): 1028-1040.e19, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28283059

RESUMEN

In eukaryotic cells, diverse stresses trigger coalescence of RNA-binding proteins into stress granules. In vitro, stress-granule-associated proteins can demix to form liquids, hydrogels, and other assemblies lacking fixed stoichiometry. Observing these phenomena has generally required conditions far removed from physiological stresses. We show that poly(A)-binding protein (Pab1 in yeast), a defining marker of stress granules, phase separates and forms hydrogels in vitro upon exposure to physiological stress conditions. Other RNA-binding proteins depend upon low-complexity regions (LCRs) or RNA for phase separation, whereas Pab1's LCR is not required for demixing, and RNA inhibits it. Based on unique evolutionary patterns, we create LCR mutations, which systematically tune its biophysical properties and Pab1 phase separation in vitro and in vivo. Mutations that impede phase separation reduce organism fitness during prolonged stress. Poly(A)-binding protein thus acts as a physiological stress sensor, exploiting phase separation to precisely mark stress onset, a broadly generalizable mechanism.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Proteínas de Unión a Poli(A)/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/fisiología , Secuencia de Aminoácidos , Gránulos Citoplasmáticos/química , Calor , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Mutagénesis , Proteínas de Unión a Poli(A)/química , Proteínas de Unión a Poli(A)/genética , Prolina/análisis , Prolina/metabolismo , Dominios Proteicos , Ribonucleasas/metabolismo , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Alineación de Secuencia , Estrés Fisiológico
14.
Mol Cell ; 84(8): 1570-1584.e7, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38537638

RESUMEN

Spatiotemporal regulation of intracellular signaling molecules, such as the 3',5'-cyclic adenosine monophosphate (cAMP)-dependent protein kinase (PKA), ensures proper cellular function. Liquid-liquid phase separation (LLPS) of the ubiquitous PKA regulatory subunit RIα promotes cAMP compartmentation and signaling specificity. However, the molecular determinants of RIα LLPS remain unclear. Here, we reveal that two separate dimerization interfaces, combined with the cAMP-induced unleashing of the PKA catalytic subunit (PKA-C) from the pseudosubstrate inhibitory sequence, drive RIα condensate formation in the cytosol of mammalian cells, which is antagonized by docking to A-kinase anchoring proteins. Strikingly, we find that the RIα pseudosubstrate region is critically involved in forming a non-canonical R:C complex, which recruits active PKA-C to RIα condensates to maintain low basal PKA activity in the cytosol. Our results suggest that RIα LLPS not only facilitates cAMP compartmentation but also spatially restrains active PKA-C, thus highlighting the functional versatility of biomolecular condensates in driving signaling specificity.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Separación de Fases , Animales , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/química , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Transducción de Señal , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Mamíferos/metabolismo
15.
Mol Cell ; 84(12): 2238-2254.e11, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38870936

RESUMEN

Transcriptional coregulators and transcription factors (TFs) contain intrinsically disordered regions (IDRs) that are critical for their association and function in gene regulation. More recently, IDRs have been shown to promote multivalent protein-protein interactions between coregulators and TFs to drive their association into condensates. By contrast, here we demonstrate how the IDR of the corepressor LSD1 excludes TF association, acting as a dynamic conformational switch that tunes repression of active cis-regulatory elements. Hydrogen-deuterium exchange shows that the LSD1 IDR interconverts between transient open and closed conformational states, the latter of which inhibits partitioning of the protein's structured domains with TF condensates. This autoinhibitory switch controls leukemic differentiation by modulating repression of active cis-regulatory elements bound by LSD1 and master hematopoietic TFs. Together, these studies unveil alternative mechanisms by which disordered regions and their dynamic crosstalk with structured regions can shape coregulator-TF interactions to control cis-regulatory landscapes and cell fate.


Asunto(s)
Elementos de Facilitación Genéticos , Histona Demetilasas , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Humanos , Proteínas Intrínsecamente Desordenadas/metabolismo , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/química , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Animales , Unión Proteica , Ratones , Diferenciación Celular , Silenciador del Gen
16.
Mol Cell ; 84(15): 2949-2965.e10, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39053456

RESUMEN

The eukaryotic nucleus has a highly organized structure. Although the spatiotemporal arrangement of spliceosomes on nascent RNA drives splicing, the nuclear architecture that directly supports this process remains unclear. Here, we show that RNA-binding proteins (RBPs) assembled on RNA form meshworks in human and mouse cells. Core and accessory RBPs in RNA splicing make two distinct meshworks adjacently but distinctly distributed throughout the nucleus. This is achieved by mutual exclusion dynamics between the charged and uncharged intrinsically disordered regions (IDRs) of RBPs. These two types of meshworks compete for spatial occupancy on pre-mRNA to regulate splicing. Furthermore, the optogenetic enhancement of the RBP meshwork causes aberrant splicing, particularly of genes involved in neurodegeneration. Genetic mutations associated with neurodegenerative diseases are often found in the IDRs of RBPs, and cells harboring these mutations exhibit impaired meshwork formation. Our results uncovered the spatial organization of RBP networks to drive RNA splicing.


Asunto(s)
Núcleo Celular , Empalme del ARN , Proteínas de Unión al ARN , Humanos , Núcleo Celular/metabolismo , Núcleo Celular/genética , Animales , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Ratones , Precursores del ARN/metabolismo , Precursores del ARN/genética , Mutación , Empalmosomas/metabolismo , Empalmosomas/genética , Células HeLa , Células HEK293
17.
Mol Cell ; 83(19): 3438-3456.e12, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37738977

RESUMEN

Transcription factors (TFs) activate enhancers to drive cell-specific gene programs in response to signals, but our understanding of enhancer assembly during signaling events is incomplete. Here, we show that androgen receptor (AR) forms condensates through multivalent interactions mediated by its N-terminal intrinsically disordered region (IDR) to orchestrate enhancer assembly in response to androgen signaling. AR IDR can be substituted by IDRs from selective proteins for AR condensation capacity and its function on enhancers. Expansion of the poly(Q) track within AR IDR results in a higher AR condensation propensity as measured by multiple methods, including live-cell single-molecule microscopy. Either weakening or strengthening AR condensation propensity impairs its heterotypic multivalent interactions with other enhancer components and diminishes its transcriptional activity. Our work reveals the requirement of an optimal level of AR condensation in mediating enhancer assembly and suggests that alteration of the fine-tuned multivalent IDR-IDR interactions might underlie AR-related human pathologies.


Asunto(s)
Elementos de Facilitación Genéticos , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Hormonas , Transducción de Señal
18.
Mol Cell ; 83(19): 3457-3469.e7, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37802023

RESUMEN

Circadian gene transcription is fundamental to metabolic physiology. Here we report that the nuclear receptor REV-ERBα, a repressive component of the molecular clock, forms circadian condensates in the nuclei of mouse liver. These condensates are dictated by an intrinsically disordered region (IDR) located in the protein's hinge region which specifically concentrates nuclear receptor corepressor 1 (NCOR1) at the genome. IDR deletion diminishes the recruitment of NCOR1 and disrupts rhythmic gene transcription in vivo. REV-ERBα condensates are located at high-order transcriptional repressive hubs in the liver genome that are highly correlated with circadian gene repression. Deletion of the IDR disrupts transcriptional repressive hubs and diminishes silencing of target genes by REV-ERBα. This work demonstrates physiological circadian protein condensates containing REV-ERBα whose IDR is required for hub formation and the control of rhythmic gene expression.


Asunto(s)
Relojes Circadianos , Ratones , Animales , Relojes Circadianos/genética , Ritmo Circadiano/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Hígado/metabolismo , Expresión Génica
19.
Annu Rev Genet ; 56: 423-439, 2022 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-36070559

RESUMEN

Human accelerated regions (HARs) are the fastest-evolving sequences in the human genome. When HARs were discovered in 2006, their function was mysterious due to scant annotation of the noncoding genome. Diverse technologies, from transgenic animals to machine learning, have consistently shown that HARs function as gene regulatory enhancers with significant enrichment in neurodevelopment. It is now possible to quantitatively measure the enhancer activity of thousands of HARs in parallel and model how each nucleotide contributes to gene expression. These strategies have revealed that many human HAR sequences function differently than their chimpanzee orthologs, though individual nucleotide changes in the same HAR may have opposite effects, consistent with compensatory substitutions. To fully evaluate the role of HARs in human evolution, it will be necessary to experimentally and computationally dissect them across more cell types and developmental stages.


Asunto(s)
Genoma Humano , Nucleótidos , Animales , Humanos , Genoma Humano/genética , Animales Modificados Genéticamente
20.
Immunity ; 54(5): 988-1001.e5, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33857421

RESUMEN

Positive selection of high-affinity B cells within germinal centers (GCs) drives affinity maturation of antibody responses. Here, we examined the mechanism underlying the parallel transition from immunoglobulin M (IgM) to IgG. Early GCs contained mostly unswitched IgM+ B cells; IgG+ B cells subsequently increased in frequency, dominating GC responses 14-21 days after antigen challenge. Somatic hypermutation and generation of high-affinity clones occurred with equal efficiency among IgM+ and IgG+ GC B cells, and inactivation of Ig class-switch recombination did not prevent depletion of IgM+ GC B cells. Instead, high-affinity IgG+ GC B cells outcompeted high-affinity IgM+ GC B cells via a selective advantage associated with IgG antigen receptor structure but independent of the extended cytoplasmic tail. Thus, two parallel forms of GC B-cell-positive selection, based on antigen receptor variable and constant regions, respectively, operate in tandem to ensure high-affinity IgG antibodies predominate in mature serum antibody responses.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Animales , Formación de Anticuerpos/inmunología , Antígenos/inmunología , Femenino , Cambio de Clase de Inmunoglobulina/inmunología , Región Variable de Inmunoglobulina/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovinos/inmunología , Hipermutación Somática de Inmunoglobulina/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA